1
|
López Garzón NA, Pinzón-Fernández MV, Saavedra T. JS, Nati-Castillo HA, Arias-Intriago M, Salazar-Santoliva C, Izquierdo-Condoy JS. Microgravity and Cellular Biology: Insights into Cellular Responses and Implications for Human Health. Int J Mol Sci 2025; 26:3058. [PMID: 40243850 PMCID: PMC11988870 DOI: 10.3390/ijms26073058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Microgravity, defined by minimal gravitational forces, represents a unique environment that profoundly influences biological systems, including human cells. This review examines the effects of microgravity on biological processes and their implications for human health. Microgravity significantly impacts the immune system by disrupting key mechanisms, such as T cell activation, cytokine production, and macrophage differentiation, leading to increased susceptibility to infections. In cancer biology, it promotes the formation of spheroids in cancer stem cells and thyroid cancer cells, which closely mimic in vivo tumor dynamics, providing novel insights for oncology research. Additionally, microgravity enhances tissue regeneration by modulating critical pathways, including Hippo and PI3K-Akt, thereby improving stem cell differentiation into hematopoietic and cardiomyocyte lineages. At the organ level, microgravity induces notable changes in hepatic metabolism, endothelial function, and bone mechanotransduction, contributing to lipid dysregulation, vascular remodeling, and accelerated bone loss. Notably, cardiomyocytes derived from human pluripotent stem cells and cultured under microgravity exhibit enhanced mitochondrial biogenesis, improved calcium handling, and advanced structural maturation, including increased sarcomere length and nuclear eccentricity. These advancements enable the development of functional cardiomyocytes, presenting promising therapeutic opportunities for treating cardiac diseases, such as myocardial infarctions. These findings underscore the dual implications of microgravity for space medicine and terrestrial health. They highlight its potential to drive advances in regenerative therapies, oncology, and immunological interventions. Continued research into the biological effects of microgravity is essential for protecting astronaut health during prolonged space missions and fostering biomedical innovations with transformative applications on Earth.
Collapse
Affiliation(s)
- Nelson Adolfo López Garzón
- Grupo de Investigación en Salud (GIS), Departamento de Medicina Interna, Universidad del Cauca, Popayan 190003, Colombia
| | | | - Jhan S. Saavedra T.
- Grupo de Investigación en Salud (GIS), Departamento de Medicina Interna, Universidad del Cauca, Popayan 190003, Colombia
| | - Humberto A. Nati-Castillo
- Interinstitutional Internal Medicine Group (GIMI 1), Department of Internal Medicine, Universidad Libre, Cali 760031, Colombia
| | | | | | | |
Collapse
|
2
|
Walker SL, Leete P, Boldison J. Tissue Resident and Infiltrating Immune Cells: Their Influence on the Demise of Beta Cells in Type 1 Diabetes. Biomolecules 2025; 15:441. [PMID: 40149976 PMCID: PMC11939886 DOI: 10.3390/biom15030441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease that results in the selective loss of pancreatic beta cells and an eventual deficit in insulin production to maintain glucose homeostasis. It is now increasingly accepted that this dynamic disease process is multifactorial; involves a variety of immune cells which contribute to an inflamed pancreatic microenvironment; and that the condition is heterogenous, resulting in variable rates of subsequent beta cell damage. In this review, we will explore the current understanding of the cellular interactions between both resident and infiltrating immune cells within the pancreatic environment, highlighting key mechanisms which may promote the beta cell destruction and islet damage associated with T1D.
Collapse
Affiliation(s)
| | | | - Joanne Boldison
- Department of Clinical and Biomedical Sciences, University of Exeter, RILD Building (Level 4), Barrack Road, Exeter EX2 5DW, UK; (S.L.W.); (P.L.)
| |
Collapse
|
3
|
Álvarez-López AI, Cruz-Chamorro I, Lardone PJ, Bejarano I, Aspiazu-Hinostroza K, Ponce-España E, Santos-Sánchez G, Álvarez-Sánchez N, Carrillo-Vico A. Melatonin, an Antitumor Necrosis Factor Therapy. J Pineal Res 2025; 77:e70025. [PMID: 39740227 DOI: 10.1111/jpi.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Tumor necrosis factor (TNF) is a biomarker of inflammation whose levels are elevated in patients with several diseases associated with dysregulation of the immune response. The main limitations of currently used anti-TNF therapies are the induction of immunodepression, which in many cases leads to serious adverse effects such as infection and cancer, and the inability to cross the blood-brain barrier in neuroinflammatory conditions. Melatonin, in addition to being a chronobiotic compound, is widely known for its antioxidant and immunomodulatory capacity to control inflammatory processes in different pathological contexts. The aim of the present review is to address human-based studies that describe the effect of melatonin on TNF production. The review includes all the articles published in PubMed databases until April 15, 2024. After depuration, 45 studies were finally included in the review, 23 related to the in vitro action of melatonin in human cells and 22 in vivo studies in humans. Most of the data reviewed support the idea that melatonin has an immunosuppressive effect on TNF levels, which, together with its low toxicity profile, low cost, and ability to cross the blood-brain barrier, points to melatonin as a potential anti-TNF therapy. Therefore, improving our knowledge of the action of melatonin in regulating TNF through appropriate clinical trials would reveal the true potential of this molecule as a possible anti-TNF therapy.
Collapse
Grants
- This work was supported by the Andalusian Government Ministry of Health PC-0019-2017, PI-0015-2018 and PEMP-0085-2020 (co-financed with FEDER funds, call Resolution of 7 July 2021 of the General Secretary for Research, Development and Innovation in Health, which calls for grants to finance research, development and innovation in biomedicine and health sciences in Andalusia by 2021), the PAIDI Program from the Andalusian Government (CTS160) and Regional Ministry of Economy and Knowledge of Andalusia (US-1263804) into the European Regional Development Fund Operational Programme 2014 to 2020. A.I.A.L. was supported by grants US-1263804 and PEMP-0085-2020. I.C.C. was supported by a postdoctoral fellowship from the Andalusian Government Ministry of Economy, Knowledge, Business, and University (DOC_00587/2020). I.B. and E.P.E were supported by the VI Program of Inner Initiative for Research and Transfer of the University of Seville [VI PPIT-US]. G.S.S. was supported by a FPU grant from the Spanish Ministerio de Educación, Cultura y Deporte (FPU16/02339). N.A.-S. was supported by a fellowship from the Andalusian Regional Ministry of Health (PC-0111-2016-0111).
Collapse
Affiliation(s)
- Ana Isabel Álvarez-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Ignacio Bejarano
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Karla Aspiazu-Hinostroza
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Universidad Católica de Cuenca, Research Department, Cuenca-Azuay, Ecuador
| | - Eduardo Ponce-España
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Guillermo Santos-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Nuria Álvarez-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
4
|
Zeger L, Barasa P, Han Y, Hellgren J, Redwan IN, Reiche ME, Florin G, Christoffersson G, Kozlova EN. Microgravity Effect on Pancreatic Islets. Cells 2024; 13:1588. [PMID: 39329769 PMCID: PMC11430520 DOI: 10.3390/cells13181588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/05/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
We previously demonstrated that boundary cap neural crest stem cells (BCs) induce the proliferation of beta-cells in vitro, increase survival of pancreatic islets (PIs) in vivo after transplantation, and themselves strongly increase their proliferation capacity after exposure to space conditions. Therefore, we asked if space conditions can induce the proliferation of beta-cells when PIs are alone or together with BCs in free-floating or 3D-printed form. During the MASER 15 sounding rocket experiment, half of the cells were exposed to 6 min of microgravity (µg), whereas another group of cells were kept in 1 g conditions in a centrifuge onboard. The proliferation marker EdU was added to the cells just before the rocket reached µg conditions. The morphological assessment revealed that PIs successfully survived and strongly proliferated, particularly in the free-floating condition, though the fusion of PIs hampered statistical analysis. Proliferation of beta-cells was displayed in 3D-printed islets two weeks after µg exposure, suggesting that the effects of µg may be delayed. Thus, PIs in 3D-printed scaffolds did not fuse, and this preparation is more suitable than free-floating specimens for morphological analysis in µg studies. PIs maintained their increased proliferation capacity for weeks after µg exposure, an effect that may not appear directly, but can emerge after a delay.
Collapse
Affiliation(s)
- Lukas Zeger
- Regenerative Neurobiology, Department of Immunology, Genetics and Pathology, Uppsala University, 75108 Uppsala, Sweden; (L.Z.); (Y.H.)
| | - Povilas Barasa
- Department of Biological Models, Institute of Biochemistry, Vilnius University, LT-08662 Vilnius, Lithuania;
| | - Yilin Han
- Regenerative Neurobiology, Department of Immunology, Genetics and Pathology, Uppsala University, 75108 Uppsala, Sweden; (L.Z.); (Y.H.)
| | - Josefin Hellgren
- CELLINK Bioprinting AB, Langfilsgatan 7, 41277 Gothenburg, Sweden; (J.H.); (I.N.R.)
| | - Itedale Namro Redwan
- CELLINK Bioprinting AB, Langfilsgatan 7, 41277 Gothenburg, Sweden; (J.H.); (I.N.R.)
| | - Myrthe E. Reiche
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, 75310 Uppsala, Sweden; (M.E.R.); (G.C.)
| | | | - Gustaf Christoffersson
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, 75310 Uppsala, Sweden; (M.E.R.); (G.C.)
| | - Elena N. Kozlova
- Regenerative Neurobiology, Department of Immunology, Genetics and Pathology, Uppsala University, 75108 Uppsala, Sweden; (L.Z.); (Y.H.)
| |
Collapse
|
5
|
Dwyer AJ, Shaheen ZR, Fife BT. Antigen-specific T cell responses in autoimmune diabetes. Front Immunol 2024; 15:1440045. [PMID: 39211046 PMCID: PMC11358097 DOI: 10.3389/fimmu.2024.1440045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Autoimmune diabetes is a disease characterized by the selective destruction of insulin-secreting β-cells of the endocrine pancreas by islet-reactive T cells. Autoimmune disease requires a complex interplay between host genetic factors and environmental triggers that promote the activation of such antigen-specific T lymphocyte responses. Given the critical involvement of self-reactive T lymphocyte in diabetes pathogenesis, understanding how these T lymphocyte populations contribute to disease is essential to develop targeted therapeutics. To this end, several key antigenic T lymphocyte epitopes have been identified and studied to understand their contributions to disease with the aim of developing effective treatment approaches for translation to the clinical setting. In this review, we discuss the role of pathogenic islet-specific T lymphocyte responses in autoimmune diabetes, the mechanisms and cell types governing autoantigen presentation, and therapeutic strategies targeting such T lymphocyte responses for the amelioration of disease.
Collapse
Affiliation(s)
- Alexander J. Dwyer
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Zachary R. Shaheen
- Center for Immunology, Department of Pediatrics, Pediatric Rheumatology, Allergy, & Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Brian T. Fife
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
6
|
Vaitaitis G, Webb T, Webb C, Sharkey C, Sharkey S, Waid D, Wagner DH. Canine diabetes mellitus demonstrates multiple markers of chronic inflammation including Th40 cell increases and elevated systemic-immune inflammation index, consistent with autoimmune dysregulation. Front Immunol 2024; 14:1319947. [PMID: 38318506 PMCID: PMC10839093 DOI: 10.3389/fimmu.2023.1319947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024] Open
Abstract
Introduction Canine diabetes mellitus (CDM) is a relatively common endocrine disease in dogs. Many CDM clinical features resemble human type 1 diabetes mellitus (T1DM), but lack of autoimmune biomarkers makes calling the disease autoimmune controversial. Autoimmune biomarkers linking CDM and T1DM would create an alternative model for drug development impacting both human and canine disease. Methods We examined peripheral blood of diagnosed CDM dog patients comparing it to healthy control (HC) dogs. Dogs were recruited to a study at the Colorado State University Veterinary Teaching Hospital and blood samples collected for blood chemistry panels, complete blood counts (CBC), and immunologic analysis. Markers of disease progression such as glycated albumin (fructosamine, the canine equivalent of human HbA1c) and c-peptide were addressed. Results Significant differences in adaptive immune lymphocytes, innate immune macrophages/monocytes and neutrophils and differences in platelets were detected between CDM and HC based on CBC. Significant differences in serum glucose, cholesterol and the liver function enzyme alkaline phosphatase were also detected. A systemic immune inflammation index (SII) and chronic inflammation index (CII) as measures of dynamic changes in adaptive and innate cells between inflammatory and non-inflammatory conditions were created with highly significant differences between CDM and HC. Th40 cells (CD4+CD40+ T cells) that are demonstrably pathogenic in mouse T1DM and able to differentiate diabetic from non-diabetic subjects in human T1DM were significantly expanded in peripheral blood mononuclear cells. Conclusions Based on each clinical finding, CDM can be categorized as an autoimmune condition. The association of significantly elevated Th40 cells in CDM when compared to HC or to osteoarthritis, a chronic but non-autoimmune disease, suggests peripheral blood Th40 cell numbers as a biomarker that reflects CDM chronic inflammation. The differences in SII and CII further underscore those findings.
Collapse
Affiliation(s)
- Gisela Vaitaitis
- Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Tracy Webb
- Department of Clinical Sciences, The Colorado State University Veterinary Teaching Hospital, Fort Collins, CO, United States
| | - Craig Webb
- Department of Clinical Sciences, The Colorado State University Veterinary Teaching Hospital, Fort Collins, CO, United States
| | - Christina Sharkey
- Department of Clinical Sciences, Montclaire Animal Clinic, Denver, CO, United States
| | - Steve Sharkey
- Department of Clinical Sciences, Montclaire Animal Clinic, Denver, CO, United States
| | - Dan Waid
- Op-T, LLC, Fitzsimmons Innovation Bioscience, Aurora, CO, United States
| | - David H. Wagner
- Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Op-T, LLC, Fitzsimmons Innovation Bioscience, Aurora, CO, United States
| |
Collapse
|
7
|
Du C, Whiddett RO, Buckle I, Chen C, Forbes JM, Fotheringham AK. Advanced Glycation End Products and Inflammation in Type 1 Diabetes Development. Cells 2022; 11:3503. [PMID: 36359899 PMCID: PMC9657002 DOI: 10.3390/cells11213503] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/18/2022] [Accepted: 10/31/2022] [Indexed: 08/08/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which the β-cells of the pancreas are attacked by the host's immune system, ultimately resulting in hyperglycemia. It is a complex multifactorial disease postulated to result from a combination of genetic and environmental factors. In parallel with increasing prevalence of T1D in genetically stable populations, highlighting an environmental component, consumption of advanced glycation end products (AGEs) commonly found in in Western diets has increased significantly over the past decades. AGEs can bind to cell surface receptors including the receptor for advanced glycation end products (RAGE). RAGE has proinflammatory roles including in host-pathogen defense, thereby influencing immune cell behavior and can activate and cause proliferation of immune cells such as islet infiltrating CD8+ and CD4+ T cells and suppress the activity of T regulatory cells, contributing to β-cell injury and hyperglycemia. Insights from studies of individuals at risk of T1D have demonstrated that progression to symptomatic onset and diagnosis can vary, ranging from months to years, providing a window of opportunity for prevention strategies. Interaction between AGEs and RAGE is believed to be a major environmental risk factor for T1D and targeting the AGE-RAGE axis may act as a potential therapeutic strategy for T1D prevention.
Collapse
Affiliation(s)
- Chenping Du
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| | - Rani O. Whiddett
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
| | - Irina Buckle
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| | - Josephine M. Forbes
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg 3084, Australia
| | - Amelia K. Fotheringham
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba 4102, Australia
- Faculty of Medicine, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
8
|
Macrophages promote growth, migration and epithelial-mesenchymal transition of renal cell carcinoma by regulating GSDMD/IL-1β axis. Cytokine 2022; 159:156021. [DOI: 10.1016/j.cyto.2022.156021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022]
|
9
|
Scherm MG, Wyatt RC, Serr I, Anz D, Richardson SJ, Daniel C. Beta cell and immune cell interactions in autoimmune type 1 diabetes: How they meet and talk to each other. Mol Metab 2022; 64:101565. [PMID: 35944899 PMCID: PMC9418549 DOI: 10.1016/j.molmet.2022.101565] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/08/2022] [Accepted: 07/27/2022] [Indexed: 10/31/2022] Open
Abstract
Background Scope of review Major conclusions
Collapse
|
10
|
Luo Z, Soläng C, Larsson R, Singh K. Interleukin-35 Prevents the Elevation of the M1/M2 Ratio of Macrophages in Experimental Type 1 Diabetes. Int J Mol Sci 2022; 23:ijms23147970. [PMID: 35887317 PMCID: PMC9320761 DOI: 10.3390/ijms23147970] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 01/03/2023] Open
Abstract
Macrophages play an important role in the early development of type 1 diabetes (T1D). Based on the phenotype, macrophages can be classified into pro-inflammatory (M1) and anti-inflammatory (M2) macrophages. Despite intensive research in the field of macrophages and T1D, the kinetic response of M1/M2 ratio has not been studied in T1D. Thus, herein, we studied the M1 and M2 macrophages in the early development of T1D using the multiple low dose streptozotocin (MLDSTZ) mouse model. We determined the proportions of M1 and M2 macrophages in thymic glands, pancreatic lymph nodes and spleens on days 3, 7 and 10 after the first injection of STZ. In addition, we investigated the effect of IL-35 in vivo on the M1/M2 ratio and IL-35+ plasmacytoid dendritic cells in diabetic mice and in vitro on the sorted macrophages. Our results revealed that the M1/M2 ratio is higher in STZ-treated mice but this was lowered upon the treatment with IL-35. Furthermore, IL-35 treated mice had lower blood glucose levels and a higher proportion of IL-35+ cells among pDCs. Macrophages treated with IL-35 in vitro also had a higher proportion of M2 macrophages. Together, our data indicate that, under diabetic conditions, pro-inflammatory macrophages increased, but IL-35 treatment decreased the pro-inflammatory macrophages and increased anti-inflammatory macrophages, further suggesting that IL-35 prevents hyperglycemia by maintaining the anti-inflammatory phenotype of macrophages and other immune cells. Thus, IL-35 should be further investigated for the treatment of T1D and other autoimmune disorders.
Collapse
|
11
|
Cano-Cano F, Gómez-Jaramillo L, Ramos-García P, Arroba AI, Aguilar-Diosdado M. IL-1β Implications in Type 1 Diabetes Mellitus Progression: Systematic Review and Meta-Analysis. J Clin Med 2022; 11:jcm11051303. [PMID: 35268394 PMCID: PMC8910979 DOI: 10.3390/jcm11051303] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 02/01/2023] Open
Abstract
During Type 1 Diabetes Mellitus (T1DM) progression, there is chronic and low-grade inflammation that could be related to the evolution of the disease. We carried out a systematic review and meta-analysis to evaluate whether peripheral levels of pro-inflammatory markers such as interleukin-1 beta (IL-1β) is significantly different among patients with or without T1DM, in gender, management of the T1DM, detection in several biological fluids, study design, age range, and glycated hemoglobin. We searched PubMed, Embase, Web of Science, and Scopus databases, and 26 relevant studies (2186 with T1DM, 2047 controls) were included. We evaluated the studies’ quality using the Newcastle−Ottawa scale. Meta-analyses were conducted, and heterogeneity and publication bias were examined. Compared with controls, IL-1β determined by immunoassays (pooled standardized mean difference (SMD): 2.45, 95% CI = 1.73 to 3.17; p < 0.001) was significantly elevated in T1DM. The compared IL-1β levels in patients <18 years (SMD = 2.81, 95% CI = 1.88−3.74) was significantly elevated. The hemoglobin-glycated (Hbg) levels in patients <18 years were compared (Hbg > 7: SMD = 5.43, 95% CI = 3.31−7.56; p = 0.001). Compared with the study design, IL-1β evaluated by ELISA (pooled SMD = 3.29, 95% CI = 2.27 to 4.30, p < 0.001) was significantly elevated in T1DM patients. IL-1β remained significantly higher in patients with a worse management of T1DM and in the early stage of T1DM. IL-1β levels determine the inflammatory environment during T1DM.
Collapse
Affiliation(s)
- Fátima Cano-Cano
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cadiz, Spain; (F.C.-C.); (L.G.-J.); (M.A.-D.)
| | - Laura Gómez-Jaramillo
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cadiz, Spain; (F.C.-C.); (L.G.-J.); (M.A.-D.)
| | - Pablo Ramos-García
- Faculty of Dentistry, University of Granada, 18011 Granada, Spain
- Correspondence: (P.R.-G.); (A.I.A.)
| | - Ana I. Arroba
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cadiz, Spain; (F.C.-C.); (L.G.-J.); (M.A.-D.)
- Department of Endocrinology and Metabolism, University Hospital Puerta del Mar, 11009 Cadiz, Spain
- Correspondence: (P.R.-G.); (A.I.A.)
| | - Manuel Aguilar-Diosdado
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, 11009 Cadiz, Spain; (F.C.-C.); (L.G.-J.); (M.A.-D.)
- Department of Endocrinology and Metabolism, University Hospital Puerta del Mar, 11009 Cadiz, Spain
| |
Collapse
|
12
|
Huang J, Pearson JA, Wong FS, Wen L, Zhou Z. Innate immunity in latent autoimmune diabetes in adults. Diabetes Metab Res Rev 2022; 38:e3480. [PMID: 34156143 PMCID: PMC8813511 DOI: 10.1002/dmrr.3480] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 11/06/2022]
Abstract
Latent autoimmune diabetes in adults (LADA) is an autoimmune disease that shares some genetic, immunological and clinical features with both type 1 diabetes and type 2 diabetes. Immune cells including CD4+ T cells, CD8+ T cells, B cells, macrophages and dendritic cells (DCs) have been detected in the pancreas of patients with LADA and a rat model of LADA. Therefore, similar to type 1 diabetes, the pathogenesis of LADA may be caused by interactions between islet β-cells and innate and adaptive immune cells. However, the role of the immunity in the initiation and progression of LADA remains largely unknown. In this review, we have summarized the potential roles of innate immunity and immune-modulators in LADA development. Furthermore, we have examined the evidence and discussed potential innate immunological reasons for the slower development of LADA compared with type 1 diabetes. More in-depth mechanistic studies are needed to fully elucidate the roles of innate immune-associated genes, molecules and cells in their contributions to LADA pathogenesis. Undertaking these studies will greatly enhance the development of new strategies and optimization of current strategies for the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Juan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Internal Medicine, Section of Endocrinology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | | | - F. Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Li Wen
- Department of Internal Medicine, Section of Endocrinology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Brodnicki TC. A Role for lncRNAs in Regulating Inflammatory and Autoimmune Responses Underlying Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:97-118. [DOI: 10.1007/978-3-030-92034-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
14
|
Amodio G, Mandelli A, Curto R, Rancoita PMV, Stabilini A, Bonfanti R, de Pellegrin M, Bosi E, Di Serio C, Battaglia M, Gregori S. Altered Frequency and Phenotype of HLA-G-Expressing DC-10 in Type 1 Diabetes Patients at Onset and in Subjects at Risk to Develop the Disease. Front Immunol 2021; 12:750162. [PMID: 34659254 PMCID: PMC8517474 DOI: 10.3389/fimmu.2021.750162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/06/2021] [Indexed: 01/21/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease resulting in progressive destruction of β-cells. Several factors affecting lymphocyte and antigen-presenting cells, including dendritic cells (DCs), contribute to defective maintenance of tolerance in T1D. DC-10 are a subset of human DCs involved in IL-10-mediated tolerance. A precise monitoring of DC-10 in the peripheral blood is possible thanks to the discovery of specific biomarkers. DC-10, being cells that naturally express HLA-G, may be used for the appropriate staging of the disease. By enumerating and phenotypically characterizing DC-10 in the peripheral blood of subjects at different stages of T1D development-first-degree relatives (FDRs) of T1D patients, without (Abneg) or with (Abpos) autoantibodies, T1D patients at onset, and age-matched healthy controls (HCs)-we showed that DC-10 contain a high proportion of HLA-G-expressing cells as compared with monocytes. We reported that a low frequency of DC-10 during disease development is paralleled with the increased proportion of pro-inflammatory cDC2 cells. Moreover, DC-10 number and phenotype differ from Abneg FDRs, Abpos FDRs, and T1D patients compared with HCs, and DC-10 from T1D patients express low levels of CD83. Finally, multiple regression analysis, considering DC-10 and HLA-G-related parameters, showed that Abneg FDRs are more similar to subjects with autoimmunity than to HCs. This is the first demonstration that impairment in DC-10 number and phenotype, specifically CD83 expression, is associated with risk of developing T1D, suggesting a possible use of CD83+ DC-10 to stratify individuals at risk of T1D in conjunction with classical prognostic factors.
Collapse
Affiliation(s)
- Giada Amodio
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Mandelli
- Immune-Mediated Diseases Unit: From Pathogenesis to Treatment, Diabetes Research Institute (DRI), Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosalia Curto
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola M. V. Rancoita
- University Center for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Angela Stabilini
- Immune-Mediated Diseases Unit: From Pathogenesis to Treatment, Diabetes Research Institute (DRI), Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Bonfanti
- Immune-Mediated Diseases Unit: From Pathogenesis to Treatment, Diabetes Research Institute (DRI), Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Pediatrics and Neonatology, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Emanuele Bosi
- Immune-Mediated Diseases Unit: From Pathogenesis to Treatment, Diabetes Research Institute (DRI), Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Internal Medicine, IRCCS San Raffaele Hospital, Milan, Italy
- TrialNet Clinical Center, IRCCS San Raffaele Hospital, Milan, Italy
| | - Clelia Di Serio
- University Center for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Manuela Battaglia
- Immune-Mediated Diseases Unit: From Pathogenesis to Treatment, Diabetes Research Institute (DRI), Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
15
|
Thomas AM, Beskid NM, Blanchfield JL, Rosado AM, García AJ, Evavold BD, Babensee JE. Localized hydrogel delivery of dendritic cells for attenuation of multiple sclerosis in a murine model. J Biomed Mater Res A 2021; 109:1247-1255. [PMID: 33040412 PMCID: PMC11250987 DOI: 10.1002/jbm.a.37118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/05/2020] [Accepted: 10/09/2020] [Indexed: 01/24/2023]
Abstract
In multiple sclerosis (MS), abnormally activated immune cells responsive to myelin proteins result in widespread damage throughout the central nervous system (CNS) and ultimately irreversible disability. Immunomodulation by delivering dendritic cells (DCs) utilizes a potent and rapid MS disease progression driver therapeutically. Here, we investigated delivering DCs for disease severity attenuation using an experimental autoimmune encephalomyelitis preclinical MS model. DCs treated with interleukin-10 (IL-10) (DC10s) were transplanted using in situ gelling poly(ethylene glycol)-based hydrogel for target site localization. DC delivery increased hydrogel longevity and altered the injection site recruited, endogenous immune cell profile within 2 days postinjection. Furthermore, hydrogel-mediated DC transplantation efficacy depended on the injection-site. DCs delivered to the neck local to MS-associated CNS-draining cervical lymph nodes attenuated paralysis, compared to untreated controls, while delivery to the flank did not alter paralysis severity. This study demonstrates that local delivery of DC10s modulates immune cell recruitment and attenuates disease progression in a preclinical model of MS.
Collapse
Affiliation(s)
- Aline M. Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
| | - Nicholas M. Beskid
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology
| | | | - Aaron M. Rosado
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
| | - Andrés J. García
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology
| | | | - Julia E. Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
| |
Collapse
|
16
|
Costa FRC, Leite JA, Rassi DM, da Silva JF, Elias-Oliveira J, Guimarães JB, Foss-Freitas MC, Câmara NOS, Pontillo A, Tostes RC, Silva JS, Carlos D. NLRP1 acts as a negative regulator of Th17 cell programming in mice and humans with autoimmune diabetes. Cell Rep 2021; 35:109176. [PMID: 34038731 DOI: 10.1016/j.celrep.2021.109176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/30/2020] [Accepted: 05/04/2021] [Indexed: 01/07/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of pancreatic β cells. We show here that the protein NOD-like receptor family pyrin domain containing 1 (NLRP1) has a key role in the pathogenesis of mouse and human T1D. More specifically, downregulation of NLRP1 expression occurs during T helper 17 (Th17) differentiation, alongside greater expression of several molecules related to Th17 cell differentiation in a signal transducers and activators of transcription 3 (STAT3)-dependent pathway. These changes lead to a consequent increase in interleukin 17 (IL-17) production within the pancreas and higher incidence of diabetes in streptozotocin (STZ)-injected mice. Finally, in patients with T1D and a SNP (rs12150220) in NLRP1, there is a robust decrease in IL-17 levels in serum and in memory Th17 cells from peripheral blood mononuclear cells. Our results demonstrate that NLRP1 acts as a negative regulator of the Th17 cell polarization program, making it an interesting target for intervention during the early stages of T1D.
Collapse
Affiliation(s)
- Frederico R C Costa
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jefferson A Leite
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Diane M Rassi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Josiane F da Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jefferson Elias-Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jhefferson B Guimarães
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria C Foss-Freitas
- Department of Clinical Medicine, Internal Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, and Caswell Diabetes Institute University of Michigan, Ann Arbor, MI, USA
| | - Niels O S Câmara
- Department of Immunology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences (ICB), University of São Paulo, São Paulo, SP, Brazil
| | - Alessandra Pontillo
- Department of Immunology, Institute of Biomedical Sciences (ICB), University of São Paulo, São Paulo, SP, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Fiocruz- Bi-Institutional Translational Medicine Platform, Ribeirão Preto, SP, Brazil
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
17
|
Xiao Y, Shu L, Wu X, Liu Y, Cheong LY, Liao B, Xiao X, Hoo RL, Zhou Z, Xu A. Fatty acid binding protein 4 promotes autoimmune diabetes by recruitment and activation of pancreatic islet macrophages. JCI Insight 2021; 6:141814. [PMID: 33690220 PMCID: PMC8119222 DOI: 10.1172/jci.insight.141814] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/18/2021] [Indexed: 12/25/2022] Open
Abstract
Both innate and adaptive immune cells are critical players in autoimmune destruction of insulin-producing β cells in type 1 diabetes. However, the early pathogenic events triggering the recruitment and activation of innate immune cells in islets remain obscure. Here we show that circulating fatty acid binding protein 4 (FABP4) level was significantly elevated in patients with type 1 diabetes and their first-degree relatives and positively correlated with the titers of several islet autoantibodies. In nonobese diabetic (NOD) mice, increased FABP4 expression in islet macrophages started from the neonatal period, well before the occurrence of overt diabetes. Furthermore, the spontaneous development of autoimmune diabetes in NOD mice was markedly reduced by pharmacological inhibition or genetic ablation of FABP4 or adoptive transfer of FABP4-deficient bone marrow cells. Mechanistically, FABP4 activated innate immune responses in islets by enhancing the infiltration and polarization of macrophages to proinflammatory M1 subtype, thus creating an inflammatory milieu required for activation of diabetogenic CD8+ T cells and shift of CD4+ helper T cells toward Th1 subtypes. These findings demonstrate FABP4 as a possible early mediator for β cell autoimmunity by facilitating crosstalk between innate and adaptive immune cells, suggesting that pharmacological inhibition of FABP4 may represent a promising therapeutic strategy for autoimmune diabetes.
Collapse
Affiliation(s)
- Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Hematologic Oncology, Cancer Center, Sun Yat-Sen University, Guangzhou, China
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Medicine, and
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Yang Liu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Medicine, and
| | - Boya Liao
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Xiaoyu Xiao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruby L.C. Hoo
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology
- Department of Medicine, and
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
Joshi K, Cameron F, Tiwari S, Mannering SI, Elefanty AG, Stanley EG. Modeling Type 1 Diabetes Using Pluripotent Stem Cell Technology. Front Endocrinol (Lausanne) 2021; 12:635662. [PMID: 33868170 PMCID: PMC8047192 DOI: 10.3389/fendo.2021.635662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/03/2021] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is increasingly being used to create in vitro models of monogenic human disorders. This is possible because, by and large, the phenotypic consequences of such genetic variants are often confined to a specific and known cell type, and the genetic variants themselves can be clearly identified and controlled for using a standardized genetic background. In contrast, complex conditions such as autoimmune Type 1 diabetes (T1D) have a polygenic inheritance and are subject to diverse environmental influences. Moreover, the potential cell types thought to contribute to disease progression are many and varied. Furthermore, as HLA matching is critical for cell-cell interactions in disease pathogenesis, any model that seeks to test the involvement of particular cell types must take this restriction into account. As such, creation of an in vitro model of T1D will require a system that is cognizant of genetic background and enables the interaction of cells representing multiple lineages to be examined in the context of the relevant environmental disease triggers. In addition, as many of the lineages critical to the development of T1D cannot be easily generated from iPSCs, such models will likely require combinations of cell types derived from in vitro and in vivo sources. In this review we imagine what an ideal in vitro model of T1D might look like and discuss how the required elements could be feasibly assembled using existing technologies. We also examine recent advances towards this goal and discuss potential uses of this technology in contributing to our understanding of the mechanisms underlying this autoimmune condition.
Collapse
Affiliation(s)
- Kriti Joshi
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences Rishikesh, Uttarakhand, India
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
| | - Fergus Cameron
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Endocrinology and Diabetes, The Royal Children’s Hospital, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, Vic, Australia
| | - Andrew G. Elefanty
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| | - Edouard G. Stanley
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| |
Collapse
|
19
|
Ke Q, Kroger CJ, Clark M, Tisch RM. Evolving Antibody Therapies for the Treatment of Type 1 Diabetes. Front Immunol 2021; 11:624568. [PMID: 33679717 PMCID: PMC7930374 DOI: 10.3389/fimmu.2020.624568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
Type 1 diabetes (T1D) is widely considered to be a T cell driven autoimmune disease resulting in reduced insulin production due to dysfunction/destruction of pancreatic β cells. Currently, there continues to be a need for immunotherapies that selectively reestablish persistent β cell-specific self-tolerance for the prevention and remission of T1D in the clinic. The utilization of monoclonal antibodies (mAb) is one strategy to target specific immune cell populations inducing autoimmune-driven pathology. Several mAb have proven to be clinically safe and exhibit varying degrees of efficacy in modulating autoimmunity, including T1D. Traditionally, mAb therapies have been used to deplete a targeted cell population regardless of antigenic specificity. However, this treatment strategy can prove detrimental resulting in the loss of acquired protective immunity. Nondepleting mAb have also been applied to modulate the function of immune effector cells. Recent studies have begun to define novel mechanisms associated with mAb-based immunotherapy that alter the function of targeted effector cell pools. These results suggest short course mAb therapies may have persistent effects for regaining and maintaining self-tolerance. Furthermore, the flexibility to manipulate mAb properties permits the development of novel strategies to target multiple antigens and/or deliver therapeutic drugs by a single mAb molecule. Here, we discuss current and potential future therapeutic mAb treatment strategies for T1D, and T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
20
|
Tootee A, Nikbin B, Ghahary A, Esfahani EN, Arjmand B, Aghayan H, Qorbani M, Larijani B. Immunopathology of Type 1 Diabetes and Immunomodulatory Effects of Stem Cells: A Narrative Review of the Literature. Endocr Metab Immune Disord Drug Targets 2021; 22:169-197. [PMID: 33538679 DOI: 10.2174/1871530321666210203212809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/11/2020] [Accepted: 10/27/2020] [Indexed: 11/22/2022]
Abstract
Type 1 Diabetes (T1D) is a complex autoimmune disorder which occurs as a result of an intricate series of pathologic interactions between pancreatic β-cells and a wide range of components of both the innate and the adaptive immune systems. Stem-cell therapy, a recently-emerged potentially therapeutic option for curative treatment of diabetes, is demonstrated to cause significant alternations to both different immune cells such as macrophages, natural killer (NK) cells, dendritic cells, T cells, and B cells and non-cellular elements including serum cytokines and different components of the complement system. Although there exists overwhelming evidence indicating that the documented therapeutic effects of stem cells on patients with T1D is primarily due to their potential for immune regulation rather than pancreatic tissue regeneration, to date, the precise underlying mechanisms remain obscure. On the other hand, immune-mediated rejection of stem cells remains one of the main obstacles to regenerative medicine. Moreover, the consequences of efferocytosis of stem-cells by the recipients' lung-resident macrophages have recently emerged as a responsible mechanism for some immune-mediated therapeutic effects of stem-cells. This review focuses on the nature of the interactions amongst different compartments of the immune systems which are involved in the pathogenesis of T1D and provides explanation as to how stem cell-based interventions can influence immune system and maintain the physiologic equilibrium.
Collapse
Affiliation(s)
- Ali Tootee
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Behrouz Nikbin
- Research Center of Molecular Immunology, Tehran University of Medical Sciences, Tehran, . Iran
| | - Aziz Ghahary
- British Columbia Professional Firefighters' Burn and Wound Healing Research Laboratory, Department of Surgery, Plastic Surgery, University of British Columbia, Vancouver, . Canada
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Babak Arjmand
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Hamidreza Aghayan
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, . Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| |
Collapse
|
21
|
Zirpel H, Roep BO. Islet-Resident Dendritic Cells and Macrophages in Type 1 Diabetes: In Search of Bigfoot's Print. Front Endocrinol (Lausanne) 2021; 12:666795. [PMID: 33912139 PMCID: PMC8072455 DOI: 10.3389/fendo.2021.666795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
The classical view of type 1 diabetes assumes that the autoimmune mediated targeting of insulin producing ß-cells is caused by an error of the immune system. Malfunction and stress of beta cells added the target tissue at the center of action. The innate immune system, and in particular islet-resident cells of the myeloid lineage, could function as a link between stressed ß-cells and activation and recognition by the adaptive immune system. We survey the role of islet-resident macrophages and dendritic cells in healthy islet homeostasis and pathophysiology of T1D. Knowledge of islet-resident antigen presenting cells in rodents is substantial, but quite scarce in humans, in particular regarding dendritic cells. Differences in blood between healthy and diseased individuals were reported, but it remains elusive to what extend these contribute to T1D onset. Increasing our understanding of the interaction between ß-cells and innate immune cells may provide new insights into disease initiation and development that could ultimately point to future treatment options. Here we review current knowledge of islet-resident macrophages and dendritic cells, place these in context of current clinical trials, and guide future research.
Collapse
|
22
|
Correlations between Iron Metabolism Parameters, Inflammatory Markers and Lipid Profile Indicators in Patients with Type 1 and Type 2 Diabetes Mellitus. J Pers Med 2020; 10:jpm10030070. [PMID: 32722394 PMCID: PMC7563504 DOI: 10.3390/jpm10030070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
This study aims to establish relationships between inflammatory status, ferrokinetics and lipid metabolism in patients with diabetes mellitus. Subclinical inflammation was assessed by levels of high-sensitive C-reactive protein, tumor necrosis factor-α and erythrocyte sedimentation rate. Iron metabolism parameters included complete blood count, serum iron, transferrin and ferritin. Metabolic status assessment included lipid profile, glycated hemoglobin and microalbuminuria measurement. As a result of the study it was possible to establish both general (universal) and diabetes mellitus (DM) type-dependent relationships between the parameters of lipid profile and metabolic control in DM. High-density lipoprotein cholesterol (HDL-C) levels negatively correlated with microalbuminuria (r = −0.293; p ˂ 0.05 for type 1 diabetes and r = −0.272; p ˂ 0.05 for type 2 diabetes). Ferritin concentration positively correlated with triglyceride level (r = 0.346; p ˂ 0.05 for type 1 diabetes and r = 0.244; p ˂ 0.05 for type 2 diabetes). In type 1 diabetes, a negative correlation was discovered between estimated glomerular filtration rate (eGFR) and LDL-C (r = −0.480; p ˂ 0.05), very low-density-lipoprotein cholesterol (VLDL-C) (r = −0.490; p ˂ 0.05) and triglycerides (r = −0.553; p ˂ 0.05), and a positive one between C-reactive protein concentration and triglyceride level (r = 0.567; p ˂ 0.05). Discovered relationships between lipid profile indices, inflammatory status and microalbuminuria confirmed mutual influence of hyperlipidemia, inflammation and nephropathy in diabetes patients. Obtained results justify the strategy of early hypolipidemic therapy in patients with diabetes mellitus to prevent the development and progression of microvascular complications.
Collapse
|
23
|
Badal D, Dayal D, Singh G, Sachdeva N. Role of DNA-LL37 complexes in the activation of plasmacytoid dendritic cells and monocytes in subjects with type 1 diabetes. Sci Rep 2020; 10:8896. [PMID: 32483133 PMCID: PMC7264208 DOI: 10.1038/s41598-020-65851-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 05/11/2020] [Indexed: 02/08/2023] Open
Abstract
Initiation of type 1 diabetes (T1D) is marked by the infiltration of plasmacytoid dendritic cells (pDCs) and monocytes in pancreatic islets. Dying beta cells release self-DNA, which forms complexes with antimicrobial peptide, LL37, and its delayed clearance can activate pDCs and monocytes. Here, we studied the phenotypic effects of DNA-LL37 complexes on pDCs and monocytes in 55 recently diagnosed T1D and 25 healthy control (HC) subjects. Following in vitro stimulation with DNA-LL37 complexes, T1D group demonstrated higher frequency and mean fluorescence intensity (MFI) of pDCs expressing IFN-α. Similarly, the monocytes in T1D group showed an increase in MFI of IFN-α. Post-stimulation, an increase in the antigen presentation and co-stimulatory ability of pDCs and monocytes was observed in T1D group, as indicated by higher expression of HLA-DR, CD80 and CD86. Upon co-culture, the stimulated monocytes and pDCs, particularly in the T1D group were able to further activate autologous CD4 + T cells, with increase in expression of CD69 and CD71. Finally, in a transwell assay, the stimulated pDCs and monocytes induced an increase in apoptosis of 1.1B4 beta cells. Additionally, we observed reduced expression of indoleamine 2,3-dioxygenase 1 (IDO1) in pDCs and monocytes of T1D subjects. Our results suggest that DNA-LL37 complexes activate pDCs and monocytes towards a proinflammatory phenotype during pathogenesis of T1D.
Collapse
Affiliation(s)
- Darshan Badal
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Devi Dayal
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Gunjan Singh
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
24
|
He W, Kapate N, Shields CW, Mitragotri S. Drug delivery to macrophages: A review of targeting drugs and drug carriers to macrophages for inflammatory diseases. Adv Drug Deliv Rev 2019; 165-166:15-40. [PMID: 31816357 DOI: 10.1016/j.addr.2019.12.001] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022]
Abstract
Macrophages play a key role in defending against foreign pathogens, healing wounds, and regulating tissue homeostasis. Driving this versatility is their phenotypic plasticity, which enables macrophages to respond to subtle cues in tightly coordinated ways. However, when this coordination is disrupted, macrophages can aid the progression of numerous diseases, including cancer, cardiovascular disease, and autoimmune disease. The central link between these disorders is aberrant macrophage polarization, which misguides their functional programs, secretory products, and regulation of the surrounding tissue microenvironment. As a result of their important and deterministic roles in both health and disease, macrophages have gained considerable attention as targets for drug delivery. Here, we discuss the role of macrophages in the initiation and progression of various inflammatory diseases, summarize the leading drugs used to regulate macrophages, and review drug delivery systems designed to target macrophages. We emphasize strategies that are approved for clinical use or are poised for clinical investigation. Finally, we provide a prospectus of the future of macrophage-targeted drug delivery systems.
Collapse
Affiliation(s)
- Wei He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Neha Kapate
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Theories about the pathogenesis of type 1 diabetes (T1D) refer to the potential of primary islet inflammatory signaling as a trigger for the loss of self-tolerance leading to disease onset. Emerging evidence suggests that extracellular vesicles (EV) may represent the missing link between inflammation and autoimmunity. Here, we review the evidence for a role of EV in the pathogenesis of T1D, as well as discuss their potential value in the clinical sphere, as biomarkers and therapeutic agents. RECENT FINDINGS EV derived from β cells are enriched in diabetogenic autoantigens and miRNAs that are selectively sorted and packaged. These EV play a pivotal role in antigen presentation and cell to cell communication leading to activation of autoimmune responses. Furthermore, recent evidence suggests the potential of EV as novel tools in clinical diagnostics and therapeutic interventions. In-depth analysis of EV cargo using modern multi-parametric technologies may be useful in enhancing our understanding of EV-mediated immune mechanisms and in identifying robust biomarkers and therapeutic strategies for T1D.
Collapse
Affiliation(s)
- Sarita Negi
- Human Islet Transplant Laboratory, Department of Surgery, D5.5736, Royal Victoria Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, T6G 2E1, Canada
| | - Alissa K Rutman
- Human Islet Transplant Laboratory, Department of Surgery, D5.5736, Royal Victoria Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, T6G 2E1, Canada
| | - Steven Paraskevas
- Human Islet Transplant Laboratory, Department of Surgery, D5.5736, Royal Victoria Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada.
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, T6G 2E1, Canada.
| |
Collapse
|
26
|
Ma WT, Gao F, Gu K, Chen DK. The Role of Monocytes and Macrophages in Autoimmune Diseases: A Comprehensive Review. Front Immunol 2019; 10:1140. [PMID: 31178867 PMCID: PMC6543461 DOI: 10.3389/fimmu.2019.01140] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
Monocytes (Mo) and macrophages (Mϕ) are key components of the innate immune system and are involved in regulation of the initiation, development, and resolution of many inflammatory disorders. In addition, these cells also play important immunoregulatory and tissue-repairing roles to decrease immune reactions and promote tissue regeneration. Several lines of evidence have suggested a causal link between the presence or activation of these cells and the development of autoimmune diseases. In addition, Mo or Mϕ infiltration in diseased tissues is a hallmark of several autoimmune diseases. However, the detailed contributions of these cells, whether they actually initiate disease or perpetuate disease progression, and whether their phenotype and functional alteration are merely epiphenomena are still unclear in many autoimmune diseases. Additionally, little is known about their heterogeneous populations in different autoimmune diseases. Elucidating the relevance of Mo and Mϕ in autoimmune diseases and the associated mechanisms could lead to the identification of more effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Wen-Tao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fei Gao
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kui Gu
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - De-Kun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
27
|
Govea-Alonso DO, Arevalo-Villalobos JI, Márquez-Escobar VA, Vimolmangkang S, Rosales-Mendoza S. An overview of tolerogenic immunotherapies based on plant-made antigens. Expert Opin Biol Ther 2019; 19:587-599. [PMID: 30892096 DOI: 10.1080/14712598.2019.1597048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Over the last two decades, genetically engineered plants became attractive and mature platforms for producing vaccines and other relevant biopharmaceuticals. Autoimmune and inflammatory disorders demand the availability of accessible treatments, and one alternative therapy is based on therapeutic vaccines able to downregulate immune responses that favor pathology progression. AREAS COVERED The current status of plant-made tolerogenic vaccines is presented with emphasis on the candidates under evaluation in test animals. Nowadays, this concept has been assessed in models of food and pollen allergies, autoimmune diabetes, asthma, arthritis, and prevention of blocking antibodies induction against a biopharmaceutical used in replacement therapies. EXPERT OPINION According to the current evidence generated at the preclinical level, plant-made tolerogenic therapies are a promise to treat several immune-related conditions, and the beginning of clinical trials is envisaged for the next decade. Advantages and limitations for this technology are discussed.
Collapse
Affiliation(s)
- Dania O Govea-Alonso
- a Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México.,b Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México
| | - Jaime I Arevalo-Villalobos
- a Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México.,b Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México
| | - Verónica A Márquez-Escobar
- a Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México.,b Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México
| | - Sornkanok Vimolmangkang
- c Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences , Chulalongkorn University , Bangkok , Thailand.,d Research Unit for Plant-Produced Pharmaceuticals , Chulalongkorn University , Bangkok , Thailand
| | - Sergio Rosales-Mendoza
- a Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México.,b Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina , Universidad Autónoma de San Luis Potosí , San Luis Potosí , México
| |
Collapse
|
28
|
Chan MWY, Viswanathan S. Recent progress on developing exogenous monocyte/macrophage-based therapies for inflammatory and degenerative diseases. Cytotherapy 2019; 21:393-415. [PMID: 30871899 DOI: 10.1016/j.jcyt.2019.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Cell-based therapies are a rapidly developing area of regenerative medicine as dynamic treatments that execute therapeutic functions multimodally. Monocytes and macrophages, as innate immune cells that control inflammation and tissue repair, are increasing popular clinical candidates due to their spectrum of functionality. In this article, we review the role of monocytes and macrophages specifically in inflammatory and degenerative disease pathology and the evidence supporting the use of these cells as an effective therapeutic strategy. We compare current strategies of exogenously polarized monocyte/macrophage therapies regarding dosage, delivery and processing to identify outcomes, advances and challenges to their clinical use. Monocytes/macrophages hold the potential to be a promising therapeutic avenue but understanding and optimization of disease-specific efficacy is needed to accelerate their clinical use.
Collapse
Affiliation(s)
- Mable Wing Yan Chan
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Sowmya Viswanathan
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Cell Therapy Program, University Health Network, Toronto, Ontario, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Rodelo-Haad C, Agüera ML, Carmona A, Navarro MD, Carracedo J, Rodriguez-Benot A, Aljama P. Pancreatic autoantibodies and CD14+CD16+ monocytes subset are associated with the impairment of ß-cell function after simultaneous pancreas-kidney transplantation. PLoS One 2019; 14:e0212547. [PMID: 30794611 PMCID: PMC6386378 DOI: 10.1371/journal.pone.0212547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/05/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic autoantibodies (AAb) has been associated with a worse pancreas graft survival after simultaneous pancreas-kidney transplantation (SPK). However, due to the variable time for AAb to become positive and the lack of early biomarkers suggesting such autoimmune activation, the mechanisms leading ß-cell destruction remain uncertain. The present study aimed to evaluate the association between post-transplant AAb and the functional impairment of the pancreatic ß-cell and also the association of such AAb with inflammation after SPK. In a longitudinal study, we analyzed the impact of post-transplant glutamic acid decarboxylase (GAD-65) and the insulinoma-associated autoantigen 2 (IA-2) AAb on pancreas graft function. Serum Hb1Ac and C-peptide (C-pep) were longitudinally compared between a group with positive posttransplant AAb (AAb+; n = 40) and another matched group with negative AAb (AAb-; n = 40) until the fifth year following seroconversion. In the cross-sectional analysis, we further evaluated the systemic signatures of inflammation by measuring pro-inflammatory CD14+CD16+ monocytes by flow-cytometry and interleukin 17-A serum levels in 38 SPK recipients and ten healthy controls. In the longitudinal study, patients with AAb+ showed higher levels of Hb1Ac (p<0.001) and lower C-pep levels (p<0.001) compared to those who remained AAb- throughout the follow-up. In the cross-sectional study, AAb+ patients showed a higher percentage of CD14+CD16+ monocytes compared with those with AAb- and the healthy controls (6.70±4.19% versus 4.0±1.84% and 3.44±0.93%; p = 0.026 and 0.009 respectively). Also, CD14+CD16+ monocytes correlated with Hb1Ac and C-pep serum levels. Multivariate logistic regression showed that posttransplant AAb+ was independently associated with a higher percentage of pro-inflammatory monocytes (adjusted-OR 1.59, 95%CI 1.05–2.40, p = 0.027). The group of patients with positive AAb also showed higher levels of IL17A as compared with the other groups (either healthy control or the negative AAb subjects). In conclusion, pancreatic AAb+ after SPK were not only associated with higher Hb1Ac and lower c-peptide serum levels but also with an increased percentage of CD14+CD16+ monocytes and higher levels of circulating IL17-A.
Collapse
Affiliation(s)
- Cristian Rodelo-Haad
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
- * E-mail:
| | - Maria Luisa Agüera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Andres Carmona
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Maria Dolores Navarro
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Julia Carracedo
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alberto Rodriguez-Benot
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Pedro Aljama
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| |
Collapse
|
30
|
Kroger CJ, Clark M, Ke Q, Tisch RM. Therapies to Suppress β Cell Autoimmunity in Type 1 Diabetes. Front Immunol 2018; 9:1891. [PMID: 30166987 PMCID: PMC6105696 DOI: 10.3389/fimmu.2018.01891] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that is generally considered to be T cell-driven. Accordingly, most strategies of immunotherapy for T1D prevention and treatment in the clinic have targeted the T cell compartment. To date, however, immunotherapy has had only limited clinical success. Although certain immunotherapies have promoted a protective effect, efficacy is often short-term and acquired immunity may be impacted. This has led to the consideration of combining different approaches with the goal of achieving a synergistic therapeutic response. In this review, we will discuss the status of various T1D therapeutic strategies tested in the clinic, as well as possible combinatorial approaches to restore β cell tolerance.
Collapse
Affiliation(s)
- Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
31
|
Weir GC, Ehlers MR, Harris KM, Kanaparthi S, Long A, Phippard D, Weiner LJ, Jepson B, McNamara JG, Koulmanda M, Strom TB. Alpha-1 antitrypsin treatment of new-onset type 1 diabetes: An open-label, phase I clinical trial (RETAIN) to assess safety and pharmacokinetics. Pediatr Diabetes 2018; 19:945-954. [PMID: 29473705 PMCID: PMC6030471 DOI: 10.1111/pedi.12660] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/07/2018] [Accepted: 02/12/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To determine the safety and pharmacokinetics of alpha-1 antitrypsin (AAT) in adults and children. RESEARCH DESIGN AND METHODS Short-term AAT treatment restores euglycemia in the non-obese mouse model of type 1 diabetes. A phase I multicenter study in 16 subjects with new-onset type 1 diabetes studied the safety and pharmacokinetics of Aralast NP (AAT). This open-label, dose-escalation study enrolled 8 adults aged 16 to 35 years and 8 children aged 8 to 15 years within 100 days of diagnosis, to receive 12 infusions of AAT: a low dose of 45 mg/kg weekly for 6 weeks, followed by a higher dose of 90 mg/kg for 6 weeks. RESULTS C-peptide secretion during a mixed meal, hemoglobin A1c (HbA1c), and insulin usage remained relatively stable during the treatment period. At 72 hours after infusion of 90 mg/kg, mean levels of AAT fell below 2.0 g/L for 7 of 15 subjects. To identify a plasma level of AAT likely to be therapeutic, pharmacodynamic ex vivo assays were performed on fresh whole blood from adult subjects. Polymerase chain reaction (PCR) analyses were performed on inhibitor of IKBKE, NOD1, TLR1, and TRAD gene expression, which are important for activation of nuclear factor-κB (NF-κB) and apoptosis pathways. AAT suppressed expression dose-dependently; 50% inhibition was achieved in the 2.5 to 5.0 mg/mL range. CONCLUSIONS AAT was well tolerated and safe in subjects with new-onset type 1 diabetes. Weekly doses of AAT greater than 90 mg/kg may be necessary for an optimal therapeutic effect.
Collapse
Affiliation(s)
- Gordon C Weir
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Mario R Ehlers
- Immune Tolerance Network, Clinical Trials Group, San Francisco, California
| | - Kristina M Harris
- Immune Tolerance Network, Biomarker & Discovery Research, Bethesda, Maryland
| | - Sai Kanaparthi
- Immune Tolerance Network, Biomarker & Discovery Research, Bethesda, Maryland
| | - Alice Long
- Benaroya Research Institute at Virginia Mason, Translational Research Program, Seattle, Western Australia
| | - Deborah Phippard
- Immune Tolerance Network, Biomarker & Discovery Research, Bethesda, Maryland
| | - Lia J Weiner
- Rho Federal Systems Division, Chapel Hill, North Carolina
| | - Brett Jepson
- Rho Federal Systems Division, Chapel Hill, North Carolina
| | - James G McNamara
- National Institute of Allergy and Infectious Diseases, Division of Allergy, Immunology and Transplant, Bethesda, Maryland
| | - Maria Koulmanda
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Terry B Strom
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Reddy S, Krogvold L, Martin C, Holland R, Choi J, Woo H, Wu F, Dahl-Jørgensen K. Distribution of IL-1β immunoreactive cells in pancreatic biopsies from living volunteers with new-onset type 1 diabetes: comparison with donors without diabetes and with longer duration of disease. Diabetologia 2018; 61:1362-1373. [PMID: 29589071 DOI: 10.1007/s00125-018-4600-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/14/2018] [Indexed: 10/17/2022]
Abstract
AIMS/HYPOTHESIS Although IL-1β is considered a key mediator of beta cell destruction, its cellular expression in islets during early type 1 diabetes remains unclear. We compared its expression in rare pancreatic biopsies from new-onset living volunteers with its expression in cadaveric pancreas sections from non-diabetic autoantibody-positive and -negative individuals and those with long-standing disease. METHODS Pancreatic biopsy sections from six new-onset living volunteers (group 1) and cadaveric sections from 13 non-diabetic autoantibody-negative donors (group 2), four non-diabetic autoantibody-positive donors (group 3) and nine donors with diabetes of longer duration (0.25-12 years of disease; group 4) were triple-immunostained for IL-1β, insulin and glucagon. Intra- and peri-islet IL-1β-positive cells in insulin-positive and -negative islets and in random exocrine fields were enumerated. RESULTS The mean number of IL-1β-positive cells per islet from each donor in peri- and intra-islet regions was <1.25 and <0.5, respectively. In all study groups, the percentage of islets with IL-1β cells in peri- and/or intra-islet regions was highly variable and ranged from 4.48% to 17.59% in group 1, 1.42% to 44.26% in group 2, 7.93% to 17.53% in group 3 and 3.85% to 42.86% in group 4, except in a single case where the value was 75%. In 25/32 donors, a higher percentage of islets showed IL-1β-positive cells in peri-islet than in intra-islet regions. In sections from diabetic donors (groups 1 and 4), a higher mean number of IL-1β-positive cells occurred in insulin-positive islets than in insulin-negative islets. In group 2, 70-90% of islets in 3/13 sections had weak-to-moderate IL-1β staining in alpha cells but staining was virtually absent or substantially reduced in the remaining groups. The mean number of exocrine IL-1β-positive cells in group 1 was lower than in the other groups. CONCLUSIONS/INTERPRETATION At onset of type 1 diabetes, the low number of islet-associated IL-1β-positive cells may be insufficient to elicit beta cell destruction. The variable expression in alpha cells in groups 2-4 suggests their cellular heterogeneity and probable physiological role. The significance of a higher but variable number of exocrine IL-1β-positive cells seen in non-diabetic individuals and those with long-term type 1 diabetes remains unclear.
Collapse
Affiliation(s)
- Shiva Reddy
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | - Lars Krogvold
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Charlton Martin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Rebecca Holland
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jaimin Choi
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Hannah Woo
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Fiona Wu
- Diabetes Unit, Auckland District Health Board, Auckland, New Zealand
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
33
|
Murai N, Ohtaki H, Watanabe J, Xu Z, Sasaki S, Yagura K, Shioda S, Nagasaka S, Honda K, Izumizaki M. Intrapancreatic injection of human bone marrow-derived mesenchymal stem/stromal cells alleviates hyperglycemia and modulates the macrophage state in streptozotocin-induced type 1 diabetic mice. PLoS One 2017; 12:e0186637. [PMID: 29073149 PMCID: PMC5657972 DOI: 10.1371/journal.pone.0186637] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 10/04/2017] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes mellitus is a progressive disease caused by the destruction of pancreatic β-cells, resulting in insulin dependency and hyperglycemia. While transplanted bone marrow-derived mesenchymal stem/stromal cells (BMMSCs) have been explored as an alternative therapeutic approach for diseases, the choice of delivery route may be a critical factor determining their sustainability. This study evaluated the effects of intrapancreatic and intravenous injection of human BMMSCs (hBMMSCs) in streptozotocin (STZ)-induced type 1 diabetic mouse model. C57/BL6 mice were intraperitoneally injected with 115 mg/kg STZ on day 0. hBMMSCs (1 × 106 cells) or vehicle were injected into the pancreas or jugular vein on day 7. Intrapancreatic, but not intravenous, hBMMSC injection significantly reduced blood glucose levels on day 28 compared with vehicle injection by the same route. This glucose-lowering effect was not induced by intrapancreatic injection of human fibroblasts as the xenograft control. Intrapancreatically injected fluorescence-labeled hBMMSCs were observed in the intra- and extra-lobular spaces of the pancreas, and intravenously injected cells were in the lung region, although the number of cells mostly decreased within 2 weeks of injection. For hBMMSCs injected twice into the pancreatic region on days 7 and 28, the injected mice had further reduced blood glucose to borderline diabetic levels on day 56. Animals injected with hBMMSCs twice exhibited increases in the plasma insulin level, number and size of islets, insulin-positive proportion of the total pancreas area, and intensity of insulin staining compared with vehicle-injected animals. We found a decrease of Iba1-positive cells in islets and an increase of CD206-positive cells in both the endocrine and exocrine pancreas. The hBMMSC injection also reduced the number of CD40-positive cells merged with glucagon immunoreactions in the islets. These results suggest that intrapancreatic injection may be a better delivery route of hBMMSCs for the treatment of type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Norimitsu Murai
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hirokazu Ohtaki
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Jun Watanabe
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
- Center for Biotechnology, Showa University, Tokyo, Japan
| | - Zhifang Xu
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Shun Sasaki
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Kazumichi Yagura
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Seiji Shioda
- Peptide Drug Innovation, Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Shoichiro Nagasaka
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
34
|
Kent SC, Mannering SI, Michels AW, Babon JAB. Deciphering the Pathogenesis of Human Type 1 Diabetes (T1D) by Interrogating T Cells from the "Scene of the Crime". Curr Diab Rep 2017; 17:95. [PMID: 28864875 PMCID: PMC5600889 DOI: 10.1007/s11892-017-0915-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Autoimmune-mediated destruction of insulin-producing β-cells within the pancreas results in type 1 diabetes (T1D), which is not yet preventable or curable. Previously, our understanding of the β-cell specific T cell repertoire was based on studies of autoreactive T cell responses in the peripheral blood of patients at risk for, or with, T1D; more recently, investigations have included immunohistochemical analysis of some T cell specificities in the pancreas from organ donors with T1D. Now, we are able to examine live, islet-infiltrating T cells from donors with T1D. RECENT FINDINGS Analysis of the T cell repertoire isolated directly from the pancreatic islets of donors with T1D revealed pro-inflammatory T cells with targets of known autoantigens, including proinsulin and glutamic acid decarboxylase, as well as modified autoantigens. We have assayed the islet-infiltrating T cell repertoire for autoreactivity and function directly from the inflamed islets of T1D organ donors. Design of durable treatments for prevention of or therapy for T1D requires understanding this repertoire.
Collapse
Affiliation(s)
- Sally C Kent
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, ASC7-2041, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria, 3065, Australia
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria, 3065, Australia
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jenny Aurielle B Babon
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, ASC7-2041, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
35
|
Helminen O, Huhta H, Kauppila JH, Lehenkari PP, Saarnio J, Karttunen TJ. Localization of nucleic acid-sensing toll-like receptors in human and mouse pancreas. APMIS 2016; 125:85-92. [DOI: 10.1111/apm.12632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/20/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Olli Helminen
- Department of Pathology; University of Oulu; Oulu Finland
- Department of Surgery; University of Oulu; Oulu Finland
- Department of Anatomy and Cell Biology; University of Oulu; Oulu Finland
- Medical Research Center Oulu; Oulu Finland
- Oulu University Hospital; Oulu Finland
| | - Heikki Huhta
- Department of Pathology; University of Oulu; Oulu Finland
- Department of Surgery; University of Oulu; Oulu Finland
- Department of Anatomy and Cell Biology; University of Oulu; Oulu Finland
- Medical Research Center Oulu; Oulu Finland
- Oulu University Hospital; Oulu Finland
| | - Joonas H. Kauppila
- Department of Pathology; University of Oulu; Oulu Finland
- Department of Surgery; University of Oulu; Oulu Finland
- Department of Anatomy and Cell Biology; University of Oulu; Oulu Finland
- Medical Research Center Oulu; Oulu Finland
- Oulu University Hospital; Oulu Finland
| | - Petri P. Lehenkari
- Department of Surgery; University of Oulu; Oulu Finland
- Department of Anatomy and Cell Biology; University of Oulu; Oulu Finland
- Medical Research Center Oulu; Oulu Finland
- Oulu University Hospital; Oulu Finland
| | - Juha Saarnio
- Department of Surgery; University of Oulu; Oulu Finland
- Medical Research Center Oulu; Oulu Finland
- Oulu University Hospital; Oulu Finland
| | - Tuomo J. Karttunen
- Department of Pathology; University of Oulu; Oulu Finland
- Medical Research Center Oulu; Oulu Finland
- Oulu University Hospital; Oulu Finland
| |
Collapse
|
36
|
Isaacs SR, Wang J, Kim KW, Yin C, Zhou L, Mi QS, Craig ME. MicroRNAs in Type 1 Diabetes: Complex Interregulation of the Immune System, β Cell Function and Viral Infections. Curr Diab Rep 2016; 16:133. [PMID: 27844276 DOI: 10.1007/s11892-016-0819-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since the discovery of the first mammalian microRNA (miRNA) more than two decades ago, a plethora of miRNAs has been identified in humans, now amounting to more than 2500. Essential for post-transcriptional regulation of gene networks integral for developmental pathways and immune response, it is not surprising that dysregulation of miRNAs is often associated with the aetiology of complex diseases including cancer, diabetes and autoimmune disorders. Despite massive expansion of small RNA studies and extensive investigation in diverse disease contexts, the role of miRNAs in type 1 diabetes has only recently been explored. Key studies using human islets have recently implicated virus-induced miRNA dysregulation as a pivotal mechanism of β cell destruction, while the interplay between miRNAs, the immune system and β cell survival has been illustrated in studies using animal and cellular models of disease. The role of specific miRNAs as major players in immune system homeostasis highlights their exciting potential as therapeutics and prognostic biomarkers of type 1 diabetes.
Collapse
Affiliation(s)
- Sonia R Isaacs
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Jie Wang
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Ki Wook Kim
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Congcong Yin
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Li Zhou
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Qing Sheng Mi
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Maria E Craig
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia.
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, 2145, Australia.
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
37
|
Yajima K, Oikawa Y, Ogata K, Hashiguchi A, Shimada A. CD4 + T cell-dominant insulitis in acute-onset Type 1 diabetes mellitus associated with intraductal papillary mucinous adenoma. Endocr J 2016; 63:841-847. [PMID: 27385564 DOI: 10.1507/endocrj.ej16-0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The loss of insulin-producing pancreatic β-cells in Type 1 diabetes mellitus (DM) is presumably the result of a T cell-mediated process. In general, CD8+ T cells are the predominant lymphocytes in the insulitis lesions, and CD4+ T cell-dominant insulitis is very rare. We present a case of a 72-year-old woman presented with excessive thirst and a 3-month history of weight loss. She was in a state of ketosis, and her plasma glucose concentration and HbA1c value were elevated. Moreover, anti-islet autoantibodies were positive, thus acute-onset Type 1 DM was diagnosed. At the time of diagnosis, a tumour was detected in the pancreas; total pancreatectomy was carried out 2 months later. The pathological diagnosis was intraductal papillary mucinous adenoma. Immunohistochemical staining of a sample of non-tumorous pancreatic tissue revealed 13 insulitis lesions infiltrated by both CD4+ and CD8+ T cells, and interestingly there were more CD4+ T cells than CD8+ T cells in the lesions. Moreover, B cells and macrophages had also infiltrated the lesions, and these two cell frequencies were both positively correlated with CD4+ as well as CD8+ T cell frequencies. This was a rare case with acute-onset Type 1 DM characterized by CD4+ T cell-dominant insulitis. Proinflammatory cytokines that can promote β-cell apoptosis or CD8+ T cell function are reported to be secreted from CD4+ T cells. Thus, together with B cells and macrophages, CD4+ T cell-associated immune responses may have, directly and/or indirectly, played a role in the pathogenesis of the Type 1 DM in this patient.
Collapse
MESH Headings
- Adenocarcinoma, Mucinous/complications
- Adenocarcinoma, Mucinous/immunology
- Age of Onset
- Aged
- Autoantibodies/blood
- CD4-Positive T-Lymphocytes/physiology
- Carcinoma, Intraductal, Noninfiltrating/complications
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Carcinoma, Pancreatic Ductal/complications
- Carcinoma, Pancreatic Ductal/immunology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/immunology
- Female
- Humans
- Insulin/blood
- Insulin/deficiency
- Islets of Langerhans/immunology
- Pancreatic Neoplasms/complications
- Pancreatic Neoplasms/immunology
Collapse
Affiliation(s)
- Ken Yajima
- Department of Internal Medicine, Federation of National Public Service Personnel Mutual Aid Associations, Tachikawa Hospital, Tachikawa 190-8531, Japan
| | | | | | | | | |
Collapse
|
38
|
Kuiper J, Moran M, Cetkovic-Cvrlje M. Exposure to polychlorinated biphenyl-153 decreases incidence of autoimmune Type 1 diabetes in non-obese diabetic mice. J Immunotoxicol 2016; 13:850-860. [DOI: 10.1080/1547691x.2016.1213333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Jordan Kuiper
- Department of Biological Sciences, St. Cloud State University, St. Cloud, MN, USA
- Laboratory for Immunology, St. Cloud State University, St. Cloud, MN, USA
| | - Michelle Moran
- Department of Biological Sciences, St. Cloud State University, St. Cloud, MN, USA
| | - Marina Cetkovic-Cvrlje
- Department of Biological Sciences, St. Cloud State University, St. Cloud, MN, USA
- Laboratory for Immunology, St. Cloud State University, St. Cloud, MN, USA
| |
Collapse
|
39
|
Wagner DH. Of the multiple mechanisms leading to type 1 diabetes, T cell receptor revision may play a prominent role (is type 1 diabetes more than a single disease?). Clin Exp Immunol 2016; 185:271-80. [PMID: 27271348 DOI: 10.1111/cei.12819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/20/2016] [Accepted: 05/31/2016] [Indexed: 12/30/2022] Open
Abstract
A single determinant factor for autoimmunity does not exist; disease development probably involves contributions from genetics, the environment and immune dysfunction. Type 1 diabetes is no exception. Genomewide-associated studies (GWAS) analysis in T1D has proved disappointing in revealing contributors to disease prediction; the only reliable marker has been human leucocyte antigen (HLA). Specific HLAs include DR3/DR4/DQ2/DQ8, for example. Because HLA molecules present antigen to T cells, it is reasonable that certain HLA molecules have a higher affinity to present self-antigen. Recent studies have shown that additional polymorphisms in HLA that are restricted to autoimmune conditions are further contributory. A caveat is that not all individuals with the appropriate 'pro-autoimmune' HLA develop an autoimmune disease. Another crucial component is autoaggressive T cells. Finding a biomarker to discriminate autoaggressive T cells has been elusive. However, a subset of CD4 helper cells that express the CD40 receptor have been described as becoming pathogenic. An interesting function of CD40 on T cells is to induce the recombination-activating gene (RAG)1/RAG2 T cell receptor recombination machinery. This observation is contrary to immunology paradigms that changes in TCR molecules cannot take place outside the thymic microenvironment. Alteration in TCR, called TCR revision, not only occurs, but may help to account for the development of autoaggressive T cells. Another interesting facet is that type 1 diabetes (T1D) may be more than a single disease; that is, multiple cellular components contribute uniquely, but result ultimately in the same clinical outcome, T1D. This review considers the process of T cell maturation and how that could favor auto-aggressive T cell development in T1D. The potential contribution of TCR revision to autoimmunity is also considered.
Collapse
Affiliation(s)
- D H Wagner
- Department of Medicine, Department of Neurology, Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
40
|
Huhta H, Helminen O, Kauppila JH, Salo T, Porvari K, Saarnio J, Lehenkari PP, Karttunen TJ. The Expression of Toll-like Receptors in Normal Human and Murine Gastrointestinal Organs and the Effect of Microbiome and Cancer. J Histochem Cytochem 2016; 64:470-82. [PMID: 27370795 PMCID: PMC4971779 DOI: 10.1369/0022155416656154] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are innate immune receptors expressed in all parts of the alimentary tract. However, analyses comparing expression in different segments and data on germ-free animals are lacking. Alimentary tract cancers show increased TLR expression. According to the field effect concept, carcinogenetic factors induce subtle cancer predisposing alterations in the whole organ. We studied TLR1 to TLR9 expression in all segments of the alimentary tract from cancer patients’ tumor-adjacent normal mucosa, healthy organ donors, and conventional and germ-free mice by using immunohistochemistry and quantitative PCR. All TLRs were expressed in all segments of the alimentary tract. Expression was most intensive in the small intestine in humans and conventional mice, but germ-free mice showed less expression in the small intestine. TLR expression levels were similar in cancer patients and organ donors. We provide systematic baseline data on the TLR expression in the alimentary tract. Normal epithelium adjacent to tumor seems to have similar TLR expression compared with healthy tissues suggesting absence of any field effect in TLR expression. Accordingly, specimens from cancer patients’ normal tumor-adjacent tissue can be used as control tissues in immunohistochemical TLR studies in gastrointestinal cancer.
Collapse
Affiliation(s)
- Heikki Huhta
- Departments of Pathology, University of Oulu, Oulu, Finland (HH, OH, JHK, KP, TJK),Surgery, University of Oulu, Oulu, Finland (HH, OH, JHK, JS, PPL),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| | - Olli Helminen
- Departments of Pathology, University of Oulu, Oulu, Finland (HH, OH, JHK, KP, TJK),Surgery, University of Oulu, Oulu, Finland (HH, OH, JHK, JS, PPL),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| | - Joonas H Kauppila
- Departments of Pathology, University of Oulu, Oulu, Finland (HH, OH, JHK, KP, TJK),Surgery, University of Oulu, Oulu, Finland (HH, OH, JHK, JS, PPL),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| | - Tuula Salo
- Dentistry, University of Oulu, Oulu, Finland (TS),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| | - Katja Porvari
- Departments of Pathology, University of Oulu, Oulu, Finland (HH, OH, JHK, KP, TJK)
| | - Juha Saarnio
- Surgery, University of Oulu, Oulu, Finland (HH, OH, JHK, JS, PPL),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| | - Petri P Lehenkari
- Surgery, University of Oulu, Oulu, Finland (HH, OH, JHK, JS, PPL),Anatomy and Cell biology, University of Oulu, Oulu, Finland (PPL),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| | - Tuomo J Karttunen
- Departments of Pathology, University of Oulu, Oulu, Finland (HH, OH, JHK, KP, TJK),Medical Research Center Oulu, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK),Oulu University Hospital, Oulu, Finland (HH, OH, JHK, TS, JS, PPL, TJK)
| |
Collapse
|
41
|
Berchtold LA, Prause M, Størling J, Mandrup-Poulsen T. Cytokines and Pancreatic β-Cell Apoptosis. Adv Clin Chem 2016; 75:99-158. [PMID: 27346618 DOI: 10.1016/bs.acc.2016.02.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery 30 years ago that inflammatory cytokines cause a concentration, activity, and time-dependent bimodal response in pancreatic β-cell function and viability has been a game-changer in the fields of research directed at understanding inflammatory regulation of β-cell function and survival and the causes of β-cell failure and destruction in diabetes. Having until then been confined to the use of pathophysiologically irrelevant β-cell toxic chemicals as a model of β-cell death, researchers could now mimic endocrine and paracrine effects of the cytokine response in vitro by titrating concentrations in the low to the high picomolar-femtomolar range and vary exposure time for up to 14-16h to reproduce the acute regulatory effects of systemic inflammation on β-cell secretory responses, with a shift to inhibition at high picomolar concentrations or more than 16h of exposure to illustrate adverse effects of local, chronic islet inflammation. Since then, numerous studies have clarified how these bimodal responses depend on discrete signaling pathways. Most interest has been devoted to the proapoptotic response dependent upon mainly nuclear factor κ B and mitogen-activated protein kinase activation, leading to gene expressional changes, endoplasmic reticulum stress, and triggering of mitochondrial dysfunction. Preclinical studies have shown preventive effects of cytokine antagonism in animal models of diabetes, and clinical trials demonstrating proof of concept are emerging. The full clinical potential of anticytokine therapies has yet to be shown by testing the incremental effects of appropriate dosing, timing, and combinations of treatments. Due to the considerable translational importance of enhancing the precision, specificity, and safety of antiinflammatory treatments of diabetes, we review here the cellular, preclinical, and clinical evidence of which of the death pathways recently proposed in the Nomenclature Committee on Cell Death 2012 Recommendations are activated by inflammatory cytokines in the pancreatic β-cell to guide the identification of antidiabetic targets. Although there are still scarce human data, the cellular and preclinical studies point to the caspase-dependent intrinsic apoptosis pathway as the prime effector of inflammatory β-cell apoptosis.
Collapse
Affiliation(s)
| | - M Prause
- University of Copenhagen, Copenhagen, Denmark
| | - J Størling
- Copenhagen Diabetes Research Center, Beta Cell Biology Group, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | |
Collapse
|
42
|
Padgett LE, Anderson B, Liu C, Ganini D, Mason RP, Piganelli JD, Mathews CE, Tse HM. Loss of NOX-Derived Superoxide Exacerbates Diabetogenic CD4 T-Cell Effector Responses in Type 1 Diabetes. Diabetes 2015; 64:4171-83. [PMID: 26269022 PMCID: PMC4657579 DOI: 10.2337/db15-0546] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) play prominent roles in numerous biological systems. While classically expressed by neutrophils and macrophages, CD4 T cells also express NADPH oxidase (NOX), the superoxide-generating multisubunit enzyme. Our laboratory recently demonstrated that superoxide-deficient nonobese diabetic (NOD.Ncf1(m1J)) mice exhibited a delay in type 1 diabetes (T1D) partially due to blunted IFN-γ synthesis by CD4 T cells. For further investigation of the roles of superoxide on CD4 T-cell diabetogenicity, the NOD.BDC-2.5.Ncf1(m1J) (BDC-2.5.Ncf1(m1J)) mouse strain was generated, possessing autoreactive CD4 T cells deficient in NOX-derived superoxide. Unlike NOD.Ncf1(m1J), stimulated BDC-2.5.Ncf1(m1J) CD4 T cells and splenocytes displayed elevated synthesis of Th1 cytokines and chemokines. Superoxide-deficient BDC-2.5 mice developed spontaneous T1D, and CD4 T cells were more diabetogenic upon adoptive transfer into NOD.Rag recipients due to a skewing toward impaired Treg suppression. Exogenous superoxide blunted exacerbated Th1 cytokines and proinflammatory chemokines to approximately wild-type levels, concomitant with reduced IL-12Rβ2 signaling and P-STAT4 (Y693) activation. These results highlight the importance of NOX-derived superoxide in curbing autoreactivity due, in part, to control of Treg function and as a redox-dependent checkpoint of effector T-cell responses. Ultimately, our studies reveal the complexities of free radicals in CD4 T-cell responses.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Brian Anderson
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Chao Liu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Douglas Ganini
- Free Radical Metabolites, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Ronald P Mason
- Free Radical Metabolites, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Jon D Piganelli
- Department of Surgery, Immunology, and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Clayton E Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| |
Collapse
|
43
|
Shi G, Sun C, Gu W, Yang M, Zhang X, Zhai N, Lu Y, Zhang Z, Shou P, Zhang Z, Ning G. Free fatty acid receptor 2, a candidate target for type 1 diabetes, induces cell apoptosis through ERK signaling. J Mol Endocrinol 2014; 53:367-80. [PMID: 25298143 DOI: 10.1530/jme-14-0065] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recent reports have highlighted the roles of free fatty acid receptor 2 (FFAR2) in the regulation of metabolic and inflammatory processes. However, the potential function of FFAR2 in type 1 diabetes (T1D) remains unexplored. Our results indicated that the mRNA level of FFAR2 was upregulated in peripheral blood mononuclear cells of T1D patients. The human FFAR2 promoter regions were cloned, and luciferase reporter assays revealed that NFκB activation induced FFAR2 expression. Furthermore, we showed that FFAR2 activation by overexpression induced cell apoptosis through ERK signaling. Finally, treatment with the FFAR2 agonists acetate or phenylacetamide 1 attenuated the inflammatory response in multiple-low-dose streptozocin-induced diabetic mice, and improved the impaired glucose tolerance. These results indicate that FFAR2 may play a protective role by inducing apoptosis of infiltrated macrophage in the pancreas through its feedback upregulation and activation, thus, in turn, improving glucose homeostasis in diabetic mice. These findings highlight FFAR2 as a potential therapeutic target of T1D, representing a link between immune response and glucose homeostasis.
Collapse
Affiliation(s)
- Guojun Shi
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Sun
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minglan Yang
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofang Zhang
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Zhai
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijian Zhang
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peishun Shou
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiguo Zhang
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrinology and MetabolismEndocrine and Metabolic E-Institutes of Shanghai Universities (EISU), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Key Laboratory for Endocrinology and Metabolism of Chinese Health Ministry, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197th Ruijin 2nd Road, Shanghai 200025, ChinaLaboratory of Endocrinology and MetabolismKey Laboratory of Stem Cell BiologyInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
In't Veld P. Insulitis in human type 1 diabetes: a comparison between patients and animal models. Semin Immunopathol 2014; 36:569-79. [PMID: 25005747 PMCID: PMC4186970 DOI: 10.1007/s00281-014-0438-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 06/24/2014] [Indexed: 01/09/2023]
Abstract
Human type 1 diabetes (T1D) is considered to be an autoimmune disease, with CD8+ T-cell-mediated cytotoxicity being directed against the insulin-producing beta cells, leading to a gradual decrease in beta cell mass and the development of chronic hyperglycemia. The histopathologically defining lesion in recent-onset T1D patients is insulitis, a relatively subtle leucocytic infiltration present in approximately 10 % of the islets of Langerhans from children with recent-onset (<1 year) disease. Due to the transient nature of the infiltrate, its heterogeneous distribution in the pancreas and the nature of the patient population, material for research is extremely rare and limited to a cumulative total of approximately 150 cases collected over the past century. Most studies on the etiopathogenesis of T1D have therefore focused on the non-obese diabetic (NOD) mouse model, which shares many genetic and immunological disease characteristics with human T1D, although its islet histopathology is remarkably different. In view of these differences and in view of the limited success of clinical immune interventions based on observations in the NOD mouse, there is a renewed focus on studying the pathogenetic process in patient material.
Collapse
Affiliation(s)
- Peter In't Veld
- Department of Pathology, Diabetes Research Center, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium,
| |
Collapse
|
45
|
Hu S, Wang J, Xu H, Wang Y, Li Z, Xue C. Fucosylated chondroitin sulphate from sea cucumber inhibits high-fat-sucrose diet-induced apoptosis in mouse pancreatic islets via down-regulating mitochondrial signaling pathway. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
46
|
Jörns A, Arndt T, Meyer zu Vilsendorf A, Klempnauer J, Wedekind D, Hedrich HJ, Marselli L, Marchetti P, Harada N, Nakaya Y, Wang GS, Scott FW, Gysemans C, Mathieu C, Lenzen S. Islet infiltration, cytokine expression and beta cell death in the NOD mouse, BB rat, Komeda rat, LEW.1AR1-iddm rat and humans with type 1 diabetes. Diabetologia 2014; 57:512-21. [PMID: 24310561 DOI: 10.1007/s00125-013-3125-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 11/08/2013] [Indexed: 12/28/2022]
Abstract
AIMS/HYPOTHESIS Research on the pathogenesis of type 1 diabetes relies heavily on good animal models. The aim of this work was to study the translational value of animal models of type 1 diabetes to the human situation. METHODS We compared the four major animal models of spontaneous type 1 diabetes, namely the NOD mouse, BioBreeding (BB) rat, Komeda rat and LEW.1AR1-iddm rat, by examining the immunohistochemistry and in situ RT-PCR of immune cell infiltrate and cytokine pattern in pancreatic islets, and by comparing findings with human data. RESULTS After type 1 diabetes manifestation CD8(+) T cells, CD68(+) macrophages and CD4(+) T cells were observed as the main immune cell types with declining frequency, in infiltrated islets of all diabetic pancreases. IL-1β and TNF-α were the main proinflammatory cytokines in the immune cell infiltrate in NOD mice, BB rats and LEW.1AR1-iddm rats, as well as in humans. The Komeda rat was the exception, with IFN-γ and TNF-α being the main cytokines. In addition, IL-17 and IL-6 and the anti-inflammatory cytokines IL-4, IL-10 and IL-13 were found in some infiltrating immune cells. Apoptotic as well as proliferating beta cells were observed in infiltrated islets. In healthy pancreases no proinflammatory cytokine expression was observed. CONCLUSIONS/INTERPRETATION With the exception of the Komeda rat, the animal models mirror very well the situation in humans with type 1 diabetes. Thus animal models of type 1 diabetes can provide meaningful information on the disease processes in the pancreas of patients with type 1 diabetes.
Collapse
Affiliation(s)
- Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, 30623, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yoshikawa A, Imagawa A, Nakata S, Fukui K, Kuroda Y, Miyata Y, Sato Y, Hanafusa T, Matsuoka TA, Kaneto H, Iwahashi H, Shimomura I. Interferon stimulated gene 15 has an anti-apoptotic effect on MIN6 cells. Endocr J 2014; 61:883-90. [PMID: 25031023 DOI: 10.1507/endocrj.ej14-0219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes, one of two major forms of diabetes, results from the complete destruction of pancreatic beta cells. Viral infection has been suggested to be a trigger of beta cell destruction, the pathogenesis of type 1 diabetes. The aim of this study was to clarify the role of the protein encoded by intherferon stimulated gene (ISG) 15, an antiviral effector, in the development of this clinical entity. We used the mouse beta cell line MIN6 to investigate the role of ISG15 and paid special attention to apoptosis. Although not detected in native MIN6 cells, free ISG15 and ISG15 conjugated proteins were both present in dose-dependently increased amounts following stimulation with interferon alpha. As assessed both by caspase 3/7 activity and an annexin V assay, the percentage of apoptotic MIN6 cells (after exposure to the inflammatory cytokines of interleukin-1beta plus interferon gamma or tumor necrosis factor alpha) was decreased by pretreatment with adenovirus-expressing ISG15 and increased by expressing a short hairpin RNA directed against ISG15. In conclusion, ISG15 has an anti-apoptotic effect on MIN6 cells. Thus, promoting ISG15 expression in the pancreatic beta cells could be a potential therapeutic approach for patients with type 1 diabetes.
Collapse
Affiliation(s)
- Atsushi Yoshikawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Morel PA. Dendritic cell subsets in type 1 diabetes: friend or foe? Front Immunol 2013; 4:415. [PMID: 24367363 PMCID: PMC3853773 DOI: 10.3389/fimmu.2013.00415] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/13/2013] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a T cell mediated autoimmune disease characterized by immune mediated destruction of the insulin-producing β cells in the islets of Langerhans. Dendritic cells (DC) have been implicated in the pathogenesis of T1D and are also used as immunotherapeutic agents. Plasmacytoid (p)DC have been shown to have both protective and pathogenic effects and a newly described merocytic DC population has been shown to break tolerance in the mouse model of T1D, the non-obese diabetic (NOD) mouse. We have used DC populations to prevent the onset of T1D in NOD mice and clinical trials of DC therapy in T1D diabetes have been initiated. In this review we will critically examine the recent published literature on the role of DC subsets in the induction and regulation of the autoimmune response in T1D.
Collapse
Affiliation(s)
- Penelope A Morel
- Department of Immunology, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
49
|
Wållberg M, Cooke A. Immune mechanisms in type 1 diabetes. Trends Immunol 2013; 34:583-91. [PMID: 24054837 DOI: 10.1016/j.it.2013.08.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 12/17/2022]
Abstract
There are three prerequisites for development of the autoimmune disease type 1 diabetes (T1D). First, β cell-reactive T cells need to be activated; second, the response needs to be proinflammatory; and finally, immune regulation of autoreactive responses must fail. Here, we describe our current understanding of the cell types and immune mechanisms involved in each of these steps leading to T1D. Novel findings regarding β cell involvement in its own destruction, the importance of the microbiota for instruction of the immune system, and recent data from studies in T1D patients are discussed. In addition, we summarise therapeutic approaches to T1D, and how these relate to the immune mechanisms involved in disease development.
Collapse
Affiliation(s)
- Maja Wållberg
- Department of Pathology, University of Cambridge, Tennis Court Rd, Cambridge CB21QP, UK.
| | | |
Collapse
|
50
|
Ghazarian L, Diana J, Beaudoin L, Larsson PG, Puri RK, van Rooijen N, Flodström-Tullberg M, Lehuen A. Protection against type 1 diabetes upon Coxsackievirus B4 infection and iNKT-cell stimulation: role of suppressive macrophages. Diabetes 2013; 62:3785-96. [PMID: 23894189 PMCID: PMC3806597 DOI: 10.2337/db12-0958] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Invariant natural killer T (iNKT) cells belong to the innate immune system and exercise a dual role as potent regulators of autoimmunity and participate in responses against different pathogens. They have been shown to prevent type 1 diabetes development and to promote antiviral responses. Many studies in the implication of environmental factors on the etiology of type 1 diabetes have suggested a link between enteroviral infections and the development of this disease. This study of the pancreatropic enterovirus Coxsackievirus B4 (CVB4) shows that although infection accelerated type 1 diabetes development in a subset of proinsulin 2-deficient NOD mice, the activation of iNKT cells by a specific agonist, α-galactosylceramide, at the time of infection inhibited the disease. Diabetes development was associated with the infiltration of pancreatic islets by inflammatory macrophages, producing high levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α and activation of anti-islet T cells. On the contrary, macrophages infiltrating the islets after CVB4 infection and iNKT-cell stimulation expressed a number of suppressive enzymes, among which indoleamine 2,3-dioxygenase was sufficient to inhibit anti-islet T-cell response and to prevent diabetes. This study highlights the critical interaction between virus and the immune system in the acceleration or prevention of type 1 diabetes.
Collapse
Affiliation(s)
- Liana Ghazarian
- INSERM U1016, Hospital Cochin/St. Vincent de Paul, Paris, France
- Université Paris Descartes and Laboratoire d’Excellence INFLAMEX, Sorbonne Paris Cité, Paris, France
| | - Julien Diana
- INSERM U1016, Hospital Cochin/St. Vincent de Paul, Paris, France
- Université Paris Descartes and Laboratoire d’Excellence INFLAMEX, Sorbonne Paris Cité, Paris, France
| | - Lucie Beaudoin
- INSERM U1016, Hospital Cochin/St. Vincent de Paul, Paris, France
- Université Paris Descartes and Laboratoire d’Excellence INFLAMEX, Sorbonne Paris Cité, Paris, France
| | - Pär G. Larsson
- Center for Infectious Medicine, Department of Medicine, the Karolinska Institute, Stockholm, Sweden
| | - Raj K. Puri
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, Maryland
| | - Nico van Rooijen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Malin Flodström-Tullberg
- Center for Infectious Medicine, Department of Medicine, the Karolinska Institute, Stockholm, Sweden
| | - Agnès Lehuen
- INSERM U1016, Hospital Cochin/St. Vincent de Paul, Paris, France
- Université Paris Descartes and Laboratoire d’Excellence INFLAMEX, Sorbonne Paris Cité, Paris, France
- Corresponding author: Agnès Lehuen,
| |
Collapse
|